1
|
Torres-Arreola AK, García OP, Estrella-Ibarra P, Campos-Maldonado F, Camacho-Barron M, Del Carmen Aburto-Fernández M, Lerma-Alvarado RM, Rodriguez-Méndez AJ, Solís-Sáinz JC, García-Solís P. Zinc Transporter ZnT1 mRNA Expression Is Negatively Associated with Leptin Serum Concentrations but Is not Associated with Insulin Resistance or Inflammatory Markers in Visceral Adipose Tissue. Biol Trace Elem Res 2024; 202:5319-5327. [PMID: 38319549 DOI: 10.1007/s12011-024-04089-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 01/28/2024] [Indexed: 02/07/2024]
Abstract
The aim of this study was to evaluate the relationship between biomarkers of chronic inflammation, insulin resistance, and zinc transporter ZnT1 expression in human visceral adipose tissue. Visceral adipose tissue obtained from 47 adults undergoing laparoscopic surgery for cholecystectomy was used to analyze ZnT1 mRNA expression by RT-qPCR. ZnT1 mRNA levels were compared between subjects with normal weight, overweight, and obesity. A significantly lower ZnT1 expression was observed in overweight and obesity compared with normal-weight subjects (p = 0.0016). Moreover, subjects with normal weight had significantly higher serum zinc concentration (97.7 ± 13.1 mg/L) than subjects with overweight (87.0 ± 12.8 mg/L) and obesity (83.1 ± 6.6 mg/L) (p = 0.002). Pearson test showed a positive correlation between serum zinc concentrations and ZnT1 mRNA expression in visceral adipose tissue (r = 0.323; p = 0.031) and a negative correlation with body mass index (r = - 0.358; p = 0.013). A linear regression model was used to analyze the associations between ZnT1 mRNA expression and serum zinc levels, insulin resistance (HOMA2-IR), serum adipokines (leptin and adiponectin), and serum inflammation biomarkers (tumor necrosis factor alpha, interleukin-6, and C-reactive protein). Interestingly, leptin concentrations were negatively associated with ZnT1 mRNA expression (p = 0.012); however, no significant associations were found for the rest of the analyzed variables. Future research is needed to analyze the causality of negative association between ZntT1 expression in visceral adipose tissue and leptin.
Collapse
Affiliation(s)
- Ana Karen Torres-Arreola
- Endocrinology and Nutrition Laboratory, Center of Advanced Biomedical Research, School of Medicine, Autonomous University of Queretaro, Carretera a Chichimequillas S/N Ejido Bolaños Av. Universidad Dirección a La Derecha, 76140, Santiago de Querétaro, Qro., Mexico
- Biomedical Sciences PhD Program, School of Medicine, Autonomous University of Queretaro, Queretaro, Mexico
| | - Olga P García
- Department of Human Nutrition, School of Natural Sciences, Autonomous University of Queretaro, Queretaro, Mexico
| | - Paulina Estrella-Ibarra
- Endocrinology and Nutrition Laboratory, Center of Advanced Biomedical Research, School of Medicine, Autonomous University of Queretaro, Carretera a Chichimequillas S/N Ejido Bolaños Av. Universidad Dirección a La Derecha, 76140, Santiago de Querétaro, Qro., Mexico
- Biological Sciences PhD Program, School of Natural Sciences, Autonomous University of Queretaro, Queretaro, Mexico
| | - Francisco Campos-Maldonado
- Endocrinology and Nutrition Laboratory, Center of Advanced Biomedical Research, School of Medicine, Autonomous University of Queretaro, Carretera a Chichimequillas S/N Ejido Bolaños Av. Universidad Dirección a La Derecha, 76140, Santiago de Querétaro, Qro., Mexico
- Biomedical Sciences PhD Program, School of Medicine, Autonomous University of Queretaro, Queretaro, Mexico
| | - Mariela Camacho-Barron
- Department of Human Nutrition, School of Natural Sciences, Autonomous University of Queretaro, Queretaro, Mexico
| | | | | | - Adriana Jheny Rodriguez-Méndez
- Endocrinology and Nutrition Laboratory, Center of Advanced Biomedical Research, School of Medicine, Autonomous University of Queretaro, Carretera a Chichimequillas S/N Ejido Bolaños Av. Universidad Dirección a La Derecha, 76140, Santiago de Querétaro, Qro., Mexico
| | - Juan Carlos Solís-Sáinz
- Endocrinology and Nutrition Laboratory, Center of Advanced Biomedical Research, School of Medicine, Autonomous University of Queretaro, Carretera a Chichimequillas S/N Ejido Bolaños Av. Universidad Dirección a La Derecha, 76140, Santiago de Querétaro, Qro., Mexico
| | - Pablo García-Solís
- Endocrinology and Nutrition Laboratory, Center of Advanced Biomedical Research, School of Medicine, Autonomous University of Queretaro, Carretera a Chichimequillas S/N Ejido Bolaños Av. Universidad Dirección a La Derecha, 76140, Santiago de Querétaro, Qro., Mexico.
| |
Collapse
|
2
|
Liu H, Li L, Lu R. ZIP transporters-regulated Zn 2+ homeostasis: A novel determinant of human diseases. J Cell Physiol 2024; 239:e31223. [PMID: 38530191 DOI: 10.1002/jcp.31223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 01/16/2024] [Accepted: 02/06/2024] [Indexed: 03/27/2024]
Abstract
As an essential trace element for organisms, zinc participates in various physiological processes, such as RNA transcription, DNA replication, cell proliferation, and cell differentiation. The destruction of zinc homeostasis is associated with various diseases. Zinc homeostasis is controlled by the cooperative action of zinc transporter proteins that are responsible for the influx and efflux of zinc. Zinc transporter proteins are mainly categorized into two families: Zrt/Irt-like protein (SLC39A/ZIP) family and zinc transporter (SLC30A/ZNT) family. ZIP transporters contain 14 members, namely ZIP1-14, which can be further divided into four subfamilies. Currently, ZIP transporters-regulated zinc homeostasis is one of the research hotspots. Cumulative evidence suggests that ZIP transporters-regulated zinc homeostasis may cause physiological dysfunction and contribute to the onset and progression of diverse diseases, such as cancers, neurological diseases, and cardiovascular diseases. In this review, we initially discuss the structure and distribution of ZIP transporters. Furthermore, we comprehensively review the latest research progress of ZIP transporters-regulated zinc homeostasis in diseases, providing a new perspective into new therapeutic targets for treating related diseases.
Collapse
Affiliation(s)
- Huimei Liu
- Department of Pharmacology, Hengyang Medical School, University of South China, Hengyang, China
| | - Lanfang Li
- Department of Pharmacology, Hengyang Medical School, University of South China, Hengyang, China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, China
| | - Ruirui Lu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
3
|
Shoji M, Ohashi T, Nagase S, Yuri H, Ichihashi K, Takagishi T, Nagata Y, Nomura Y, Fukunaka A, Kenjou S, Miyake H, Hara T, Yoshigai E, Fujitani Y, Sakurai H, Dos Santos HG, Fukada T, Kuzuhara T. Possible involvement of zinc transporter ZIP13 in myogenic differentiation. Sci Rep 2024; 14:8052. [PMID: 38609428 PMCID: PMC11014994 DOI: 10.1038/s41598-024-56912-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 03/12/2024] [Indexed: 04/14/2024] Open
Abstract
Ehlers-Danlos syndrome spondylodysplastic type 3 (EDSSPD3, OMIM 612350) is an inherited recessive connective tissue disorder that is caused by loss of function of SLC39A13/ZIP13, a zinc transporter belonging to the Slc39a/ZIP family. We previously reported that patients with EDSSPD3 harboring a homozygous loss of function mutation (c.221G > A, p.G64D) in ZIP13 exon 2 (ZIP13G64D) suffer from impaired development of bone and connective tissues, and muscular hypotonia. However, whether ZIP13 participates in the early differentiation of these cell types remains unclear. In the present study, we investigated the role of ZIP13 in myogenic differentiation using a murine myoblast cell line (C2C12) as well as patient-derived induced pluripotent stem cells (iPSCs). We found that ZIP13 gene expression was upregulated by myogenic stimulation in C2C12 cells, and its knockdown disrupted myotubular differentiation. Myocytes differentiated from iPSCs derived from patients with EDSSPD3 (EDSSPD3-iPSCs) also exhibited incomplete myogenic differentiation. Such phenotypic abnormalities of EDSSPD3-iPSC-derived myocytes were corrected by genomic editing of the pathogenic ZIP13G64D mutation. Collectively, our findings suggest the possible involvement of ZIP13 in myogenic differentiation, and that EDSSPD3-iPSCs established herein may be a promising tool to study the molecular basis underlying the clinical features caused by loss of ZIP13 function.
Collapse
Affiliation(s)
- Masaki Shoji
- Laboratory of Biochemistry, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Nishihamahouji, Yamashirocho, Tokushima-City, Tokushima, 770-8514, Japan.
| | - Takuto Ohashi
- Laboratory of Molecular and Cellular Physiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Nishihamahouji, Yamashirocho, Tokushima-City, Tokushima, 770-8514, Japan
| | - Saki Nagase
- Laboratory of Biochemistry, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Nishihamahouji, Yamashirocho, Tokushima-City, Tokushima, 770-8514, Japan
| | - Haato Yuri
- Laboratory of Biochemistry, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Nishihamahouji, Yamashirocho, Tokushima-City, Tokushima, 770-8514, Japan
| | - Kenta Ichihashi
- Laboratory of Biochemistry, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Nishihamahouji, Yamashirocho, Tokushima-City, Tokushima, 770-8514, Japan
| | - Teruhisa Takagishi
- Laboratory of Molecular and Cellular Physiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Nishihamahouji, Yamashirocho, Tokushima-City, Tokushima, 770-8514, Japan
| | - Yuji Nagata
- Laboratory of Molecular and Cellular Physiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Nishihamahouji, Yamashirocho, Tokushima-City, Tokushima, 770-8514, Japan
| | - Yuki Nomura
- Laboratory of Molecular and Cellular Physiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Nishihamahouji, Yamashirocho, Tokushima-City, Tokushima, 770-8514, Japan
| | - Ayako Fukunaka
- Laboratory of Developmental Biology and Metabolism, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi-City, Gunma, Japan
| | - Sae Kenjou
- Laboratory of Molecular and Cellular Physiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Nishihamahouji, Yamashirocho, Tokushima-City, Tokushima, 770-8514, Japan
| | - Hatsuna Miyake
- Laboratory of Molecular and Cellular Physiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Nishihamahouji, Yamashirocho, Tokushima-City, Tokushima, 770-8514, Japan
| | - Takafumi Hara
- Laboratory of Molecular and Cellular Physiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Nishihamahouji, Yamashirocho, Tokushima-City, Tokushima, 770-8514, Japan
| | - Emi Yoshigai
- Laboratory of Molecular and Cellular Physiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Nishihamahouji, Yamashirocho, Tokushima-City, Tokushima, 770-8514, Japan
| | - Yoshio Fujitani
- Laboratory of Developmental Biology and Metabolism, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi-City, Gunma, Japan
| | - Hidetoshi Sakurai
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto-City, Kyoto, Japan
| | | | - Toshiyuki Fukada
- Laboratory of Molecular and Cellular Physiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Nishihamahouji, Yamashirocho, Tokushima-City, Tokushima, 770-8514, Japan.
| | - Takashi Kuzuhara
- Laboratory of Biochemistry, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Nishihamahouji, Yamashirocho, Tokushima-City, Tokushima, 770-8514, Japan.
| |
Collapse
|
4
|
Franco C, Canzoniero LMT. Zinc homeostasis and redox alterations in obesity. Front Endocrinol (Lausanne) 2024; 14:1273177. [PMID: 38260166 PMCID: PMC10800374 DOI: 10.3389/fendo.2023.1273177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 12/18/2023] [Indexed: 01/24/2024] Open
Abstract
Impairment of both cellular zinc and redox homeostasis is a feature of several chronic diseases, including obesity. A significant two-way interaction exists between redox metabolism and the relatively redox-inert zinc ion. Redox metabolism critically influences zinc homeostasis and controls its cellular availability for various cellular functions by regulating zinc exchange from/to zinc-binding proteins. Zinc can regulate redox metabolism and exhibits multiple pro-antioxidant properties. On the other hand, even minor disturbances in zinc status and zinc homeostasis affect systemic and cellular redox homeostasis. At the cellular level, zinc homeostasis is regulated by a multi-layered machinery consisting of zinc-binding molecules, zinc sensors, and two selective families of zinc transporters, the Zinc Transporter (ZnT) and Zrt, Irt-like protein (ZIP). In the present review, we summarize the current state of knowledge on the role of the mutual interaction between zinc and redox homeostasis in physiology and pathophysiology, pointing to the role of zinc in the alterations responsible for redox stress in obesity. Since zinc transporters primarily control zinc homeostasis, we describe how changes in the expression and activity of these zinc-regulating proteins are associated with obesity.
Collapse
|
5
|
Chen B, Yu P, Chan WN, Xie F, Zhang Y, Liang L, Leung KT, Lo KW, Yu J, Tse GMK, Kang W, To KF. Cellular zinc metabolism and zinc signaling: from biological functions to diseases and therapeutic targets. Signal Transduct Target Ther 2024; 9:6. [PMID: 38169461 PMCID: PMC10761908 DOI: 10.1038/s41392-023-01679-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 09/15/2023] [Accepted: 10/10/2023] [Indexed: 01/05/2024] Open
Abstract
Zinc metabolism at the cellular level is critical for many biological processes in the body. A key observation is the disruption of cellular homeostasis, often coinciding with disease progression. As an essential factor in maintaining cellular equilibrium, cellular zinc has been increasingly spotlighted in the context of disease development. Extensive research suggests zinc's involvement in promoting malignancy and invasion in cancer cells, despite its low tissue concentration. This has led to a growing body of literature investigating zinc's cellular metabolism, particularly the functions of zinc transporters and storage mechanisms during cancer progression. Zinc transportation is under the control of two major transporter families: SLC30 (ZnT) for the excretion of zinc and SLC39 (ZIP) for the zinc intake. Additionally, the storage of this essential element is predominantly mediated by metallothioneins (MTs). This review consolidates knowledge on the critical functions of cellular zinc signaling and underscores potential molecular pathways linking zinc metabolism to disease progression, with a special focus on cancer. We also compile a summary of clinical trials involving zinc ions. Given the main localization of zinc transporters at the cell membrane, the potential for targeted therapies, including small molecules and monoclonal antibodies, offers promising avenues for future exploration.
Collapse
Affiliation(s)
- Bonan Chen
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
- CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Peiyao Yu
- Department of Pathology, Nanfang Hospital and Basic Medical College, Southern Medical University, Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Wai Nok Chan
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
- CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Fuda Xie
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
- CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Yigan Zhang
- Institute of Biomedical Research, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Li Liang
- Department of Pathology, Nanfang Hospital and Basic Medical College, Southern Medical University, Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Kam Tong Leung
- Department of Pediatrics, The Chinese University of Hong Kong, Hong Kong, China
| | - Kwok Wai Lo
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Jun Yu
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Gary M K Tse
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Wei Kang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China.
- CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China.
| | - Ka Fai To
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
6
|
Kwon J, Yeh YS, Kawarasaki S, Minamino H, Fujita Y, Okamatsu-Ogura Y, Takahashi H, Nomura W, Matsumura S, Yu R, Kimura K, Saito M, Inagaki N, Inoue K, Kawada T, Goto T. Mevalonate biosynthesis pathway regulates the development and survival of brown adipocytes. iScience 2023; 26:106161. [PMID: 36895651 PMCID: PMC9988578 DOI: 10.1016/j.isci.2023.106161] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 01/08/2023] [Accepted: 02/03/2023] [Indexed: 02/11/2023] Open
Abstract
The high thermogenic activity of brown adipose tissue (BAT) has received considerable attention. Here, we demonstrated the role of the mevalonate (MVA) biosynthesis pathway in the regulation of brown adipocyte development and survival. The inhibition of 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR), the rate-limiting enzyme in the MVA pathway and the molecular target of statins, suppressed brown adipocyte differentiation by suppressing protein geranylgeranylation-mediated mitotic clonal expansion. The development of BAT in neonatal mice exposed to statins during the fetal period was severely impaired. Moreover, statin-induced geranylgeranyl pyrophosphate (GGPP) deficiency led to the apoptosis of mature brown adipocytes. Brown adipocyte-specific Hmgcr knockout induced BAT atrophy and disrupted thermogenesis. Importantly, both genetic and pharmacological inhibition of HMGCR in adult mice induced morphological changes in BAT accompanied by an increase in apoptosis, and statin-treated diabetic mice showed worsened hyperglycemia. These findings revealed that MVA pathway-generated GGPP is indispensable for BAT development and survival.
Collapse
Affiliation(s)
- Jungin Kwon
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto 611-0011, Japan
| | - Yu-Sheng Yeh
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto 611-0011, Japan
| | - Satoko Kawarasaki
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto 611-0011, Japan
| | - Hiroto Minamino
- Department of Diabetes, Endocrinology, and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Yoshihito Fujita
- Department of Diabetes, Endocrinology, and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Yuko Okamatsu-Ogura
- Departments of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Haruya Takahashi
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto 611-0011, Japan
| | - Wataru Nomura
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto 611-0011, Japan.,Research Unit for Physiological Chemistry, the Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto 606-8501, Japan
| | - Shigenobu Matsumura
- Division of Clinical Nutrition, Graduate School of Comprehensive Rehabilitation, Osaka Prefecture University, Osaka 583-0872, Japan
| | - Rina Yu
- Department of Food Science and Nutrition, University of Ulsan, Ulsan 44610, Republic of Korea
| | - Kazuhiro Kimura
- Departments of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Masayuki Saito
- Departments of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Nobuya Inagaki
- Department of Diabetes, Endocrinology, and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Kazuo Inoue
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto 611-0011, Japan.,Research Unit for Physiological Chemistry, the Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto 606-8501, Japan
| | - Teruo Kawada
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto 611-0011, Japan.,Research Unit for Physiological Chemistry, the Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto 606-8501, Japan
| | - Tsuyoshi Goto
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto 611-0011, Japan.,Research Unit for Physiological Chemistry, the Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto 606-8501, Japan
| |
Collapse
|
7
|
Jiang J, Zhou D, Zhang A, Yu W, Du L, Yuan H, Zhang C, Wang Z, Jia X, Zhang ZN, Luan B. Thermogenic adipocyte-derived zinc promotes sympathetic innervation in male mice. Nat Metab 2023; 5:481-494. [PMID: 36879120 DOI: 10.1038/s42255-023-00751-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 01/31/2023] [Indexed: 03/08/2023]
Abstract
Sympathetic neurons activate thermogenic adipocytes through release of catecholamine; however, the regulation of sympathetic innervation by thermogenic adipocytes is unclear. Here, we identify primary zinc ion (Zn) as a thermogenic adipocyte-secreted factor that promotes sympathetic innervation and thermogenesis in brown adipose tissue and subcutaneous white adipose tissue in male mice. Depleting thermogenic adipocytes or antagonizing β3-adrenergic receptor on adipocytes impairs sympathetic innervation. In obesity, inflammation-induced upregulation of Zn chaperone protein metallothionein-2 decreases Zn secretion from thermogenic adipocytes and leads to decreased energy expenditure. Furthermore, Zn supplementation ameliorates obesity by promoting sympathetic neuron-induced thermogenesis, while sympathetic denervation abrogates this antiobesity effect. Thus, we have identified a positive feedback mechanism for the reciprocal regulation of thermogenic adipocytes and sympathetic neurons. This mechanism is important for adaptive thermogenesis and could serve as a potential target for the treatment of obesity.
Collapse
Affiliation(s)
- Junkun Jiang
- Department of Endocrinology, Tongji Hospital Affiliated to Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Donglei Zhou
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Anke Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Wenjing Yu
- Department of Endocrinology, Tongji Hospital Affiliated to Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Lei Du
- Department of Metabolic Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Huiwen Yuan
- Department of Endocrinology, Tongji Hospital Affiliated to Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Chuan Zhang
- Department of Endocrinology, Tongji Hospital Affiliated to Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Zelin Wang
- Department of Endocrinology, Tongji Hospital Affiliated to Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Xuyang Jia
- Department of Metabolic Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhen-Ning Zhang
- Translational Medical Center for Stem Cell Therapy & Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Bing Luan
- Department of Endocrinology, Tongji Hospital Affiliated to Tongji University School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
8
|
Zinc and Zinc Transporters in Dermatology. Int J Mol Sci 2022; 23:ijms232416165. [PMID: 36555806 PMCID: PMC9785331 DOI: 10.3390/ijms232416165] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/09/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Zinc is an important trace mineral in the human body and a daily intake of zinc is required to maintain a healthy status. Over the past decades, zinc has been used in formulating topical and systemic therapies for various skin disorders owing to its wound healing and antimicrobial properties. Zinc transporters play a major role in maintaining the integrity of the integumentary system by controlling zinc homeostasis within dermal layers. Mutations and abnormal function of zinc-transporting proteins can lead to disease development, such as spondylocheirodysplastic Ehlers-Danlos syndrome (SCD-EDS) and acrodermatitis enteropathica (AE) which can be fatal if left untreated. This review discusses the layers of the skin, the importance of zinc and zinc transporters in each layer, and the various skin disorders caused by zinc deficiency, in addition to zinc-containing compounds used for treating different skin disorders and skin protection.
Collapse
|
9
|
Islam T, Albracht-Schulte K, Ramalingam L, Schlabritz-Lutsevich N, Park OH, Zabet-Moghaddam M, Kalupahana NS, Moustaid-Moussa N. Anti-inflammatory mechanisms of polyphenols in adipose tissue: role of gut microbiota, intestinal barrier integrity and zinc homeostasis. J Nutr Biochem 2022; 115:109242. [PMID: 36442715 DOI: 10.1016/j.jnutbio.2022.109242] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 09/18/2022] [Accepted: 10/14/2022] [Indexed: 11/27/2022]
Abstract
Obesity is associated with an imbalance of micro-and macro-nutrients, gut dysbiosis, and a "leaky" gut phenomenon. Polyphenols, such as curcumin, resveratrol, and anthocyanins may alleviate the systemic effects of obesity, potentially by improving gut microbiota, intestinal barrier integrity (IBI), and zinc homeostasis. The essential micronutrient zinc plays a crucial role in the regulation of enzymatic processes, including inflammation, maintenance of the microbial ecology, and intestinal barrier integrity. In this review, we focus on IBI- which prevents intestinal lipopolysaccharide (LPS) leakage - as a critical player in polyphenol-mediated protective effects against obesity-associated white adipose tissue (WAT) inflammation. This occurs through mechanisms that block the movement of the bacterial endotoxin LPS across the gut barrier. Available research suggests that polyphenols reduce WAT and systemic inflammation via crosstalk with inflammatory NF-κB, the mammalian target of rapamycin (mTOR) signaling and zinc homeostasis.
Collapse
Affiliation(s)
- Tariful Islam
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas, USA; Obesity Research Institute, Texas Tech University, Lubbock, Texas, USA
| | - Kembra Albracht-Schulte
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas, USA; Obesity Research Institute, Texas Tech University, Lubbock, Texas, USA
| | - Latha Ramalingam
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas, USA; Obesity Research Institute, Texas Tech University, Lubbock, Texas, USA
| | - Natalia Schlabritz-Lutsevich
- Obesity Research Institute, Texas Tech University, Lubbock, Texas, USA; Advanced Fertility Center, Odessa, Texas, USA
| | - Oak-Hee Park
- Obesity Research Institute, Texas Tech University, Lubbock, Texas, USA; College of Human Sciences, Texas Tech University, Lubbock, Texas, USA
| | - Masoud Zabet-Moghaddam
- Obesity Research Institute, Texas Tech University, Lubbock, Texas, USA; Center for Biotechnology and Genomics, Texas Tech University, Lubbock, Texas, USA
| | - Nishan S Kalupahana
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas, USA; Obesity Research Institute, Texas Tech University, Lubbock, Texas, USA; Department of Physiology, University of Peradeniya, Peradeniya, Sri Lanka
| | - Naima Moustaid-Moussa
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas, USA; Obesity Research Institute, Texas Tech University, Lubbock, Texas, USA.
| |
Collapse
|
10
|
Liu SZ, Xu YC, Tan XY, Zhao T, Zhang DG, Yang H, Luo Z. Transcriptional Regulation and Protein Localization of Zip10, Zip13 and Zip14 Transporters of Freshwater Teleost Yellow Catfish Pelteobagrus fulvidraco Following Zn Exposure in a Heterologous HEK293T Model. Int J Mol Sci 2022; 23:8034. [PMID: 35887381 PMCID: PMC9321221 DOI: 10.3390/ijms23148034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 07/15/2022] [Accepted: 07/18/2022] [Indexed: 12/04/2022] Open
Abstract
Zip family proteins are involved in the control of zinc (Zn) ion homeostasis. The present study cloned the promoters and investigated the transcription responses and protein subcellular localizations of three LIV-1 subfamily members (zip10, zip13, and zip14) from common freshwater teleost yellow catfish, Pelteobagrus fulvidraco, using in vitro cultured HEK293T model cells. The 2278 bp, 1917 bp, and 1989 bp sequences of zip10, zip13, and zip14 promoters, respectively, were subcloned into pGL3-Basic plasmid for promoter activity analysis. The pcDNA3.1 plasmid coding EGFP tagged pfZip10, pfZip13, and pfZip14 were generated for subsequent confocal microscope analysis. Several potential transcription factors' binding sites were predicted within the promoters. In vitro promoter analysis in the HEK293T cells showed that high Zn administration significantly reduced the transcriptional activities of the zip10, zip13, and zip14 promoters. The -2017 bp/-2004 bp MRE in the zip10 promoter, the -360 bp/-345 bp MRE in the zip13 promoter, and the -1457 bp/-1442 bp MRE in the zip14 promoter were functional loci that were involved in the regulation of the three zips. The -606 bp/-594 bp KLF4 binding site in the zip13 promoter was a functional locus responsible for zinc-responsive regulation of zip13. The -1383 bp/-1375 bp STAT3 binding site in the zip14 promoter was a functional locus responsible for zinc-responsive regulation of zip14. Moreover, confocal microscope analysis indicated that zinc incubation significantly reduced the fluorescence intensity of pfZip10-EGFP and pfZip14-EGFP but had no significant influence on pfZip13-EGFP fluorescence intensity. Further investigation found that pfZip10 localizes on cell membranes, pfZip14 colocalized with both cell membranes and lysosome, and pfZip13 colocalized with intracellular ER and Golgi. Our research illustrated the transcription regulation of zip10, zip13, and zip14 from P. fulvidraco under zinc administration, which provided a reference value for the mechanisms involved in Zip-family-mediated control of zinc homeostasis in vertebrates.
Collapse
Affiliation(s)
- Sheng-Zan Liu
- Hubei Hongshan Laboratory, Fishery College, Huazhong Agricultural University, Wuhan 430070, China; (S.-Z.L.); (Y.-C.X.); (X.-Y.T.); (T.Z.); (D.-G.Z.); (H.Y.)
| | - Yi-Chuang Xu
- Hubei Hongshan Laboratory, Fishery College, Huazhong Agricultural University, Wuhan 430070, China; (S.-Z.L.); (Y.-C.X.); (X.-Y.T.); (T.Z.); (D.-G.Z.); (H.Y.)
| | - Xiao-Ying Tan
- Hubei Hongshan Laboratory, Fishery College, Huazhong Agricultural University, Wuhan 430070, China; (S.-Z.L.); (Y.-C.X.); (X.-Y.T.); (T.Z.); (D.-G.Z.); (H.Y.)
| | - Tao Zhao
- Hubei Hongshan Laboratory, Fishery College, Huazhong Agricultural University, Wuhan 430070, China; (S.-Z.L.); (Y.-C.X.); (X.-Y.T.); (T.Z.); (D.-G.Z.); (H.Y.)
| | - Dian-Guang Zhang
- Hubei Hongshan Laboratory, Fishery College, Huazhong Agricultural University, Wuhan 430070, China; (S.-Z.L.); (Y.-C.X.); (X.-Y.T.); (T.Z.); (D.-G.Z.); (H.Y.)
| | - Hong Yang
- Hubei Hongshan Laboratory, Fishery College, Huazhong Agricultural University, Wuhan 430070, China; (S.-Z.L.); (Y.-C.X.); (X.-Y.T.); (T.Z.); (D.-G.Z.); (H.Y.)
| | - Zhi Luo
- Hubei Hongshan Laboratory, Fishery College, Huazhong Agricultural University, Wuhan 430070, China; (S.-Z.L.); (Y.-C.X.); (X.-Y.T.); (T.Z.); (D.-G.Z.); (H.Y.)
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China
| |
Collapse
|
11
|
Chen CW, Chen LK, Huang TY, Yang DM, Liu SY, Tsai PJ, Chen TH, Lin HF, Juan CC. Nitric Oxide Mobilizes Intracellular Zn2+ via the GC/cGMP/PKG Signaling Pathway and Stimulates Adipocyte Differentiation. Int J Mol Sci 2022; 23:ijms23105488. [PMID: 35628299 PMCID: PMC9143299 DOI: 10.3390/ijms23105488] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/03/2022] [Accepted: 05/11/2022] [Indexed: 12/10/2022] Open
Abstract
Plasma and tissue zinc ion levels are associated with the development of obesity. Previous studies have suggested that zinc ions may regulate adipocyte metabolism and that nitric oxide (NO) plays a pivotal role in the regulation of adipocyte physiology. Our previous study showed that chronic NO deficiency causes a significant decrease in adipose tissue mass in rats. Studies also suggested that zinc ions play an important modulatory role in regulating NO function. This study aims to explore the role of zinc ions in NO-regulated adipocyte differentiation. We hypothesized that NO could increase intracellular Zn2+ level and then stimulate adipocyte differentiation. ZnCl2 and the NO donor, NONOate, were used to explore the effects of Zn2+ and NO on adipocyte differentiation. Regulatory mechanisms of NO on intracellular Zn2+ mobilization were determined by detection. Then, Zn2+-selective chelator TPEN was used to clarify the role of intracellular Zn2+ on NO-regulated adipocyte differentiation. Furthermore, the relationship between adipocyte size, Zn2+ level, and NOS expression in human subcutaneous fat tissue was elucidated. Results showed that both ZnCl2 and NO stimulated adipocyte differentiation in a dose-dependent manner. NO stimulated intracellular Zn2+ mobilization in adipocytes through the guanylate cyclase (GC)/cyclic guanosine monophosphate (cGMP)/protein kinase G (PKG) pathway, and NO-stimulated adipocyte differentiation was Zn2+-dependent. In human subcutaneous adipose tissue, adipocyte size was negatively correlated with expression of eNOS. In conclusion, NO treatment stimulates intracellular Zn2+ mobilization through the GC/cGMP/PKG pathway, subsequently stimulating adipocyte differentiation.
Collapse
Affiliation(s)
- Chien-Wei Chen
- College of Human Development and Health, National Taipei University of Nursing and Health Sciences, Taipei 112303, Taiwan;
| | - Luen-Kui Chen
- Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan; (L.-K.C.); (T.-Y.H.); (S.-Y.L.)
| | - Tai-Ying Huang
- Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan; (L.-K.C.); (T.-Y.H.); (S.-Y.L.)
| | - De-Ming Yang
- Institute of Biophotonics, College of Biomedical Science and Engineering, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan;
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| | - Shui-Yu Liu
- Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan; (L.-K.C.); (T.-Y.H.); (S.-Y.L.)
| | - Pei-Jiun Tsai
- Institute of Anatomy and Cell Biology, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan; (P.-J.T.); (T.-H.C.)
- Department of Critical Care Medicine, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Trauma Center, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| | - Tien-Hua Chen
- Institute of Anatomy and Cell Biology, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan; (P.-J.T.); (T.-H.C.)
- Trauma Center, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Division of General Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| | - Heng-Fu Lin
- Division of Trauma, Department of Surgery, Far-Eastern Memorial Hospital, New Taipei City 220216, Taiwan
- Graduate Institute of Medicine, Yuan Ze University, Taoyuan 320315, Taiwan
- Correspondence: (H.-F.L.); (C.-C.J.)
| | - Chi-Chang Juan
- Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan; (L.-K.C.); (T.-Y.H.); (S.-Y.L.)
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Department of Education and Research, Taipei City Hospital, Taipei 103212, Taiwan
- Correspondence: (H.-F.L.); (C.-C.J.)
| |
Collapse
|
12
|
Sophisticated expression responses of ZNT1 and MT in response to changes in the expression of ZIPs. Sci Rep 2022; 12:7334. [PMID: 35513474 PMCID: PMC9072671 DOI: 10.1038/s41598-022-10925-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 04/15/2022] [Indexed: 12/16/2022] Open
Abstract
The zinc homeostatic proteins Zn transporter 1 (ZNT1) and metallothionein (MT) function in dampening increases in cytosolic zinc concentrations. Conversely, the expression of ZNT1 and MT is expected to be suppressed during decreases in cytosolic zinc concentrations. Thus, ZNT1/MT homeostatic responses are considered to be essential for maintaining cellular zinc homeostasis because cellular zinc concentrations are readily altered by changes in the expression of several Zrt-/Irt-like proteins (ZIPs) under both physiological and pathological conditions. However, this notion remains to be tested experimentally. Here, we investigated the aforementioned homeostatic process by analyzing ZNT1 and MT protein expression in response to ZIP expression. Overexpression of cell-surface-localized ZIPs, such as ZIP4 and ZIP5, increased the cellular zinc content, which caused an increase in the expression of cell-surface ZNT1 and cytosolic MT in the absence of zinc supplementation in the culture medium. By contrast, elimination of the overexpressed ZIP4 and ZIP5 resulted in decreased expression of ZNT1 but not MT, which suggests that differential regulation of ZNT1 and MT expression at the protein level underlies the homeostatic responses necessary for zinc metabolism under certain conditions. Moreover, increased expression of apically localized ZIP4 facilitated basolateral ZNT1 expression in polarized cells, which indicates that such a coordinated expression mechanism is crucial for vectorial transcellular transport. Our results provide novel insights into the physiological maintenance of cellular zinc homeostasis in response to alterations in cytosolic zinc concentrations caused by changes in the expression of ZIPs.
Collapse
|
13
|
Hara T, Yoshigai E, Ohashi T, Fukada T. Zinc transporters as potential therapeutic targets: An updated review. J Pharmacol Sci 2022; 148:221-228. [PMID: 35063137 DOI: 10.1016/j.jphs.2021.11.007] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 11/13/2021] [Accepted: 11/22/2021] [Indexed: 12/28/2022] Open
Abstract
Zinc is an essential trace element that plays important roles in the regulation of various physiological responses in the body. Zinc deficiency is known to cause various health problems, including dysgeusia, skin disorders, and immune disorders. Therefore, the maintenance of healthy zinc content in the body is critical to our healthy life. Zinc homeostasis is tightly controlled by two of the solute carrier protein families SLC30A and SLC39A, called zinc transporters. In the last decade, research on zinc biology has made dramatic progress based on the physiological and functional analysis of zinc transporters in the fields of molecular biology, human genetics, and drug discovery. In particular, since the association between zinc transporters and human diseases was recently reported using human genetics and gene knockout mouse studies, zinc and zinc signals controlled by zinc transporters have been considered useful therapeutic targets. In this review, we introduce the importance of zinc homeostasis based on the findings of zinc transporter functions and their signals in relation to human diseases.
Collapse
Affiliation(s)
- Takafumi Hara
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, Japan
| | - Emi Yoshigai
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, Japan
| | - Takuto Ohashi
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, Japan
| | - Toshiyuki Fukada
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, Japan.
| |
Collapse
|
14
|
Lack of TRPV1 aggravates obesity-associated hypertension through the disturbance of mitochondrial Ca2+ homeostasis in brown adipose tissue. Hypertens Res 2022; 45:789-801. [PMID: 35043013 PMCID: PMC9010289 DOI: 10.1038/s41440-021-00842-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 11/09/2021] [Accepted: 11/30/2021] [Indexed: 11/23/2022]
Abstract
The combination of obesity and hypertension is associated with high morbidity and mortality; however, the mechanism underlying obesity-induced hypertension remains unclear. In this study, we detected the possible effects of TRPV1, a previously identified antihypertensive calcium (Ca2+) channel in adipose tissue, on the occurrence of obesity and hypertension in mice lacking UCP1, a spontaneously genetically manipulated obesity model, by generating TRPV1 and UCP1 double knockout mice. In these mice, obesity and hypertension appeared earlier and were more severe than in mice with the knockout of UCP1 or TRPV1 alone. The knockout of TRPV1 in UCP1 knockout mice further reduced functional brown adipose tissue (BAT) generation; decreased resting oxygen consumption, heat production, and locomotor activities; and was accompanied by severe mitochondrial respiratory dysfunction in BAT. Mechanistically, TRPV1, UCP1, and LETM1 acted as a complex to maintain an appropriate mitochondrial Ca2+ level, and TRPV1 knockout caused a compensatory increase in mitochondrial Ca2+ uptake via LETM1 activation. However, the compensatory response was blocked in UCP1−/− mice, resulting in dramatically reduced mitochondrial Ca2+ uptake and higher production of ATP and oxidative stress. This study provides in vivo evidence for the critical role of BAT mitochondrial Ca2+ homeostasis in obesity-associated hypertension and indicates that the TRPV1/UCP1/LETM1 complex may be an alternative intervention target.
Collapse
|
15
|
Tamura Y. The Role of Zinc Homeostasis in the Prevention of Diabetes Mellitus and Cardiovascular Diseases. J Atheroscler Thromb 2021; 28:1109-1122. [PMID: 34148917 PMCID: PMC8592709 DOI: 10.5551/jat.rv17057] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 05/25/2021] [Indexed: 11/30/2022] Open
Abstract
Zinc is an essential micronutrient for human health and is involved in various biological functions, such as growth, metabolism, and immune function. In recent years, research on intracellular zinc dynamics has progressed, and it has become clear that zinc transporters strictly control intracellular zinc localization, zinc regulates the functions of various proteins and signal transduction pathways as a second messenger similar to calcium ions, and intracellular zinc dyshomeostasis is associated with impaired insulin synthesis, secretion, sensitivity, lipid metabolism, and vascular function. Numerous animal and human studies have shown that zinc deficiency may be associated with the risk factors for diabetes and cardiovascular diseases (CVDs) and zinc administration might be beneficial for the prevention and treatment of these diseases. Therefore, an understanding of zinc biology may help the establishment of novel strategies for the prevention and treatment of diabetes and CVDs. This review will summarize the current knowledge on the role of zinc homeostasis in the pathogenesis of diabetes and atherosclerosis and will discuss the potential of zinc in the prevention of these diseases.
Collapse
Affiliation(s)
- Yukinori Tamura
- Division of Physiology and Biochemistry, Faculty of Nutrition, Kobe Gakuin University, Kobe, Japan
| |
Collapse
|
16
|
Vroman R, Malfait AM, Miller RE, Malfait F, Syx D. Animal Models of Ehlers-Danlos Syndromes: Phenotype, Pathogenesis, and Translational Potential. Front Genet 2021; 12:726474. [PMID: 34712265 PMCID: PMC8547655 DOI: 10.3389/fgene.2021.726474] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 08/10/2021] [Indexed: 01/09/2023] Open
Abstract
The Ehlers-Danlos syndromes (EDS) are a group of heritable connective tissues disorders mainly characterized by skin hyperextensibility, joint hypermobility and generalized tissue fragility. Currently, 14 EDS subtypes each with particular phenotypic features are recognized and are caused by genetic defects in 20 different genes. All of these genes are involved in the biosynthesis and/or fibrillogenesis of collagens at some level. Although great progress has been made in elucidating the molecular basis of different EDS subtypes, the pathogenic mechanisms underlying the observed phenotypes remain poorly understood, and consequentially, adequate treatment and management options for these conditions remain scarce. To date, several animal models, mainly mice and zebrafish, have been described with defects in 14 of the 20 hitherto known EDS-associated genes. These models have been instrumental in discerning the functions and roles of the corresponding proteins during development, maturation and repair and in portraying their roles during collagen biosynthesis and/or fibrillogenesis, for some even before their contribution to an EDS phenotype was elucidated. Additionally, extensive phenotypical characterization of these models has shown that they largely phenocopy their human counterparts, with recapitulation of several clinical hallmarks of the corresponding EDS subtype, including dermatological, cardiovascular, musculoskeletal and ocular features, as well as biomechanical and ultrastructural similarities in tissues. In this narrative review, we provide a comprehensive overview of animal models manifesting phenotypes that mimic EDS with a focus on engineered mouse and zebrafish models, and their relevance in past and future EDS research. Additionally, we briefly discuss domestic animals with naturally occurring EDS phenotypes. Collectively, these animal models have only started to reveal glimpses into the pathophysiological aspects associated with EDS and will undoubtably continue to play critical roles in EDS research due to their tremendous potential for pinpointing (common) signaling pathways, unveiling possible therapeutic targets and providing opportunities for preclinical therapeutic interventions.
Collapse
Affiliation(s)
- Robin Vroman
- Center for Medical Genetics, Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Anne-Marie Malfait
- Division of Rheumatology, Rush University Medical Center, Chicago, IL, United States
| | - Rachel E. Miller
- Division of Rheumatology, Rush University Medical Center, Chicago, IL, United States
| | - Fransiska Malfait
- Center for Medical Genetics, Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Delfien Syx
- Center for Medical Genetics, Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| |
Collapse
|
17
|
Abstract
Since the discovery of manifest Zn deficiency in 1961, the increasing number of studies demonstrated the association between altered Zn status and multiple diseases. In this chapter, we provide a review of the most recent advances on the role of Zn in health and disease (2010-20), with a special focus on the role of Zn in neurodegenerative and neurodevelopmental disorders, diabetes and obesity, male and female reproduction, as well as COVID-19. In parallel with the revealed tight association between ASD risk and severity and Zn status, the particular mechanisms linking Zn2+ and ASD pathogenesis like modulation of synaptic plasticity through ProSAP/Shank scaffold, neurotransmitter metabolism, and gut microbiota, have been elucidated. The increasing body of data indicate the potential involvement of Zn2+ metabolism in neurodegeneration. Systemic Zn levels in Alzheimer's and Parkinson's disease were found to be reduced, whereas its sequestration in brain may result in modulation of amyloid β and α-synuclein processing with subsequent toxic effects. Zn2+ was shown to possess adipotropic effects through the role of zinc transporters, zinc finger proteins, and Zn-α2-glycoprotein in adipose tissue physiology, underlying its particular role in pathogenesis of obesity and diabetes mellitus type 2. Recent findings also contribute to further understanding of the role of Zn2+ in spermatogenesis and sperm functioning, as well as oocyte development and fertilization. Finally, Zn2+ was shown to be the potential adjuvant therapy in management of novel coronavirus infection (COVID-19), underlining the perspectives of zinc in management of old and new threats.
Collapse
Affiliation(s)
- Anatoly V Skalny
- IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia; Yaroslavl State University, Yaroslavl, Russia
| | - Michael Aschner
- IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia; Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Alexey A Tinkov
- IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia; Yaroslavl State University, Yaroslavl, Russia.
| |
Collapse
|
18
|
Converging vulnerability factors for compulsive food and drug use. Neuropharmacology 2021; 196:108556. [PMID: 33862029 DOI: 10.1016/j.neuropharm.2021.108556] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 03/29/2021] [Accepted: 04/03/2021] [Indexed: 12/12/2022]
Abstract
Highly palatable foods and substance of abuse have intersecting neurobiological, metabolic and behavioral effects relevant for understanding vulnerability to conditions related to food (e.g., obesity, binge eating disorder) and drug (e.g., substance use disorder) misuse. Here, we review data from animal models, clinical populations and epidemiological evidence in behavioral, genetic, pathophysiologic and therapeutic domains. Results suggest that consumption of highly palatable food and drugs of abuse both impact and conversely are regulated by metabolic hormones and metabolic status. Palatable foods high in fat and/or sugar can elicit adaptation in brain reward and withdrawal circuitry akin to substances of abuse. Intake of or withdrawal from palatable food can impact behavioral sensitivity to drugs of abuse and vice versa. A robust literature suggests common substrates and roles for negative reinforcement, negative affect, negative urgency, and impulse control deficits, with both highly palatable foods and substances of abuse. Candidate genetic risk loci shared by obesity and alcohol use disorders have been identified in molecules classically associated with both metabolic and motivational functions. Finally, certain drugs may have overlapping therapeutic potential to treat obesity, diabetes, binge-related eating disorders and substance use disorders. Taken together, data are consistent with the hypotheses that compulsive food and substance use share overlapping, interacting substrates at neurobiological and metabolic levels and that motivated behavior associated with feeding or substance use might constitute vulnerability factors for one another. This article is part of the special issue on 'Vulnerabilities to Substance Abuse'.
Collapse
|
19
|
Wang J, Cheng X, Zhao H, Yang Q, Xu Z. Downregulation of the zinc transporter SLC39A13 (ZIP13) is responsible for the activation of CaMKII at reperfusion and leads to myocardial ischemia/reperfusion injury in mouse hearts. J Mol Cell Cardiol 2020; 152:69-79. [PMID: 33307093 DOI: 10.1016/j.yjmcc.2020.12.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 12/03/2020] [Accepted: 12/04/2020] [Indexed: 12/11/2022]
Abstract
While Zn2+ dyshomeostasis is known to contribute to ischemia/reperfusion (I/R) injury, the roles of zinc transporters that are responsible for Zn2+ homeostasis in the pathogenesis of I/R injury remain to be addressed. This study reports that ZIP13 (SLC39A13), a zinc transporter, plays a role in myocardial I/R injury by modulating the Ca2+ signaling pathway rather than by regulating Zn2+ transport. ZIP13 is downregulated upon reperfusion in mouse hearts or in H9c2 cells at reoxygenation. Ca2+ but not Zn2+ was responsible for ZIP13 downregulation, implying that ZIP13 may play a role in I/R injury through the Ca2+ signaling pathway. In line with our assumption, knockout of ZIP13 resulted in phosphorylation (Thr287) of Ca2+-calmodulin-dependent protein kinase (CaMKII), indicating that downregulation of ZIP13 leads to CaMKII activation. Further studies showed that the heart-specific knockout of ZIP13 enhanced I/R-induced CaMKII phosphorylation in mouse hearts. In contrast, overexpression of ZIP13 suppressed I/R-induced CaMKII phosphorylation. Moreover, the heart-specific knockout of ZIP13 exacerbated myocardial infarction in mouse hearts subjected to I/R, whereas overexpression of ZIP13 reduced infarct size. In addition, knockout of ZIP13 induced increases of mitochondrial Ca2+, ROS, mitochondrial swelling, decrease in the mitochondrial respiration control rate (RCR), and dissipation of mitochondrial membrane potential (ΔΨm) in a CaMKII-dependent manner. These data suggest that downregulation of ZIP13 at reperfusion contributes to myocardial I/R injury through activation of CaMKII and the mitochondrial death pathway.
Collapse
Affiliation(s)
- Jie Wang
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin 300070, China
| | - Xinxin Cheng
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin 300070, China
| | - Huanhuan Zhao
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin 300070, China
| | - Qing Yang
- Department of Cardiology, General Hospital, Tianjin Medical University, Tianjin 300052, China
| | - Zhelong Xu
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin 300070, China; Department of Cardiology, General Hospital, Tianjin Medical University, Tianjin 300052, China.
| |
Collapse
|
20
|
The role of labile Zn 2+ and Zn 2+-transporters in the pathophysiology of mitochondria dysfunction in cardiomyocytes. Mol Cell Biochem 2020; 476:971-989. [PMID: 33225416 DOI: 10.1007/s11010-020-03964-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 10/23/2020] [Indexed: 02/06/2023]
Abstract
An important energy supplier of cardiomyocytes is mitochondria, similar to other mammalian cells. Studies have demonstrated that any defect in the normal processes controlled by mitochondria can lead to abnormal ROS production, thereby high oxidative stress as well as lack of ATP. Taken into consideration, the relationship between mitochondrial dysfunction and overproduction of ROS as well as the relation between increased ROS and high-level release of intracellular labile Zn2+, those bring into consideration the importance of the events related with those stimuli in cardiomyocytes responsible from cellular Zn2+-homeostasis and responsible Zn2+-transporters associated with the Zn2+-homeostasis and Zn2+-signaling. Zn2+-signaling, controlled by cellular Zn2+-homeostatic mechanisms, is regulated with intracellular labile Zn2+ levels, which are controlled, especially, with the two Zn2+-transporter families; ZIPs and ZnTs. Our experimental studies in mammalian cardiomyocytes and human heart tissue showed that Zn2+-transporters localizes to mitochondria besides sarco(endo)plasmic reticulum and Golgi under physiological condition. The protein levels as well as functions of those transporters can re-distribute under pathological conditions, therefore, they can interplay among organelles in cardiomyocytes to adjust a proper intracellular labile Zn2+ level. In the present review, we aimed to summarize the already known Zn2+-transporters localize to mitochondria and function to stabilize not only the cellular Zn2+ level but also cellular oxidative stress status. In conclusion, one can propose that a detailed understanding of cellular Zn2+-homeostasis and Zn2+-signaling through mitochondria may emphasize the importance of new mitochondria-targeting agents for prevention and/or therapy of cardiovascular dysfunction in humans.
Collapse
|
21
|
Rodríguez A, Catalán V, Ramírez B, Unamuno X, Portincasa P, Gómez-Ambrosi J, Frühbeck G, Becerril S. Impact of adipokines and myokines on fat browning. J Physiol Biochem 2020; 76:227-240. [PMID: 32236810 DOI: 10.1007/s13105-020-00736-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 02/24/2020] [Indexed: 02/07/2023]
Abstract
Since the discovery of leptin in 1994, the adipose tissue (AT) is not just considered a passive fat storage organ but also an extremely active secretory and endocrine organ that secretes a large variety of hormones, called adipokines, involved in energy metabolism. Adipokines may not only contribute to AT dysfunction and obesity, but also in fat browning, a process that induces a phenotypic switch from energy-storing white adipocytes to thermogenic brown fat-like cells. The fat browning process and, consequently, thermogenesis can also be stimulated by physical exercise. Contracting skeletal muscle is a metabolically active tissue that participates in several endocrine functions through the production of bioactive factors, collectively termed myokines, proposed as the mediators of physical activity-induced health benefits. Myokines affect muscle mass, have profound effects on glucose and lipid metabolism, and promote browning and thermogenesis of white AT in an endocrine and/or paracrine manner. The present review focuses on the role of different myokines and adipokines in the regulation of fat browning, as well as in the potential cross-talk between AT and skeletal muscle, in order to control body weight, energy expenditure and thermogenesis.
Collapse
Affiliation(s)
- A Rodríguez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Avda. Pío XII, 36, 31008, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Sevilla, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - V Catalán
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Avda. Pío XII, 36, 31008, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Sevilla, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - B Ramírez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Avda. Pío XII, 36, 31008, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Sevilla, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - X Unamuno
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Avda. Pío XII, 36, 31008, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Sevilla, Spain
- Medical Engineering Laboratory, University of Navarra, Pamplona, Spain
| | - P Portincasa
- Clinica Medica "A. Murri", Department of Biomedical Sciences and Human Oncology, Policlinico Hospital, University of Bari Medical School, 70124, Bari, Italy
| | - J Gómez-Ambrosi
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Avda. Pío XII, 36, 31008, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Sevilla, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - G Frühbeck
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Avda. Pío XII, 36, 31008, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Sevilla, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - Sara Becerril
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Avda. Pío XII, 36, 31008, Pamplona, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Sevilla, Spain.
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain.
| |
Collapse
|
22
|
Brito S, Lee MG, Bin BH, Lee JS. Zinc and Its Transporters in Epigenetics. Mol Cells 2020; 43:323-330. [PMID: 32274919 PMCID: PMC7191051 DOI: 10.14348/molcells.2020.0026] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 03/25/2020] [Accepted: 04/04/2020] [Indexed: 12/19/2022] Open
Abstract
Epigenetic events like DNA methylation and histone modification can alter heritable phenotypes. Zinc is required for the activity of various epigenetic enzymes, such as DNA methyltransferases (DNMTs), histone acetyltransferases (HATs), histone deacetylases (HDACs), and histone demethylases, which possess several zinc binding sites. Thus, the dysregulation of zinc homeostasis can lead to epigenetic alterations. Zinc homeostasis is regulated by Zinc Transporters (ZnTs), Zrt- and Irt-like proteins (ZIPs), and the zinc storage protein metallothionein (MT). Recent advances revealed that ZIPs modulate epigenetics. ZIP10 deficiency was found to result in reduced HATs, confirming its involvement in histone acetylation for rigid skin barrier formation. ZIP13 deficiency, which is associated with Spondylocheirodysplastic Ehlers-Danlos syndrome (SCD-EDS), increases DNMT activity, leading to dysgenesis of dermis via improper gene expressions. However, the precise molecular mechanisms remain to be elucidated. Future molecular studies investigating the involvement of zinc and its transporters in epigenetics are warranted.
Collapse
Affiliation(s)
- Sofia Brito
- Department of Biological Sciences, Ajou University, Suwon 6499, Korea
- These authors contributed equally to this work
| | - Mi-Gi Lee
- Bio-Center, Gyeonggido Business and Science Accelerator, Suwon 169, Korea
- These authors contributed equally to this work
| | - Bum-Ho Bin
- Department of Biological Sciences, Ajou University, Suwon 6499, Korea
| | - Jong-Soo Lee
- Department of Biological Sciences, Ajou University, Suwon 6499, Korea
| |
Collapse
|
23
|
Chen Z, Gordillo-Martinez F, Jiang L, He P, Hong W, Wei X, Staines KA, Macrae VE, Zhang C, Yu D, Fu X, Zhu D. Zinc ameliorates human aortic valve calcification through GPR39 mediated ERK1/2 signalling pathway. Cardiovasc Res 2020; 117:820-835. [PMID: 32259211 DOI: 10.1093/cvr/cvaa090] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 02/11/2020] [Accepted: 04/01/2020] [Indexed: 12/12/2022] Open
Abstract
AIMS Calcific aortic valve disease (CAVD) is the most common heart valve disease in the Western world. It has been reported that zinc is accumulated in calcified human aortic valves. However, whether zinc directly regulates CAVD is yet to be elucidated. The present study sought to determine the potential role of zinc in the pathogenesis of CAVD. METHODS AND RESULTS Using a combination of a human valve interstitial cell (hVIC) calcification model, human aortic valve tissues, and blood samples, we report that 20 μM zinc supplementation attenuates hVIC in vitro calcification, and that this is mediated through inhibition of apoptosis and osteogenic differentiation via the zinc-sensing receptor GPR39-dependent ERK1/2 signalling pathway. Furthermore, we report that GPR39 protein expression is dramatically reduced in calcified human aortic valves, and there is a significant reduction in zinc serum levels in patients with CAVD. Moreover, we reveal that 20 μM zinc treatment prevents the reduction of GPR39 observed in calcified hVICs. We also show that the zinc transporter ZIP13 and ZIP14 are significantly increased in hVICs in response to zinc treatment. Knockdown of ZIP13 or ZIP14 significantly inhibited hVIC in vitro calcification and osteogenic differentiation. CONCLUSIONS Together, these findings suggest that zinc is a novel inhibitor of CAVD, and report that zinc transporter ZIP13 and ZIP14 are important regulators of hVIC in vitro calcification and osteogenic differentiation. Zinc supplementation may offer a potential therapeutic strategy for CAVD.
Collapse
Affiliation(s)
- Ziying Chen
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510260, China
| | - Flora Gordillo-Martinez
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510260, China
| | - Lei Jiang
- Guangdong Geriatric Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Pengcheng He
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510100, China
| | - Wanzi Hong
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510100, China
| | - Xuebiao Wei
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510100, China
| | - Katherine A Staines
- School of Applied Sciences, Edinburgh Napier University, Edinburgh EH11 4BN, UK
| | - Vicky E Macrae
- The Roslin Institute, RDSVS, Easter Bush Campus, University of Edinburgh, Midlothian EH25 9RG, UK
| | - Chunxiang Zhang
- Department of Biomedical Engineering, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Danqing Yu
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510100, China
| | - Xiaodong Fu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510260, China
| | - Dongxing Zhu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510260, China
| |
Collapse
|
24
|
Abdollahi S, Toupchian O, Jayedi A, Meyre D, Tam V, Soltani S. Zinc Supplementation and Body Weight: A Systematic Review and Dose-Response Meta-analysis of Randomized Controlled Trials. Adv Nutr 2020; 11:398-411. [PMID: 31504083 PMCID: PMC7442320 DOI: 10.1093/advances/nmz084] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 05/07/2019] [Accepted: 07/17/2019] [Indexed: 12/24/2022] Open
Abstract
The aim of this study was to determine the effect of zinc supplementation on anthropometric measures. In this systematic review and dose-response meta-analysis, we searched PubMed, Scopus, ISI Web of Science, and the Cochrane Library from database inception to August 2018 for relevant randomized controlled trials. Mean differences and SDs for each outcome were pooled using a random-effects model. Furthermore, a dose-response analysis for zinc dosage was performed using a fractional polynomial model. Quality of evidence was evaluated using the Grading of Recommendations Assessment, Development and Evaluation (GRADE) methodology. Twenty-seven trials (n = 1438 participants) were included in the meta-analysis. There were no significant changes in anthropometric measures after zinc supplementation in the overall analysis. However, subgroup analyses revealed that zinc supplementation increased body weight in individuals undergoing hemodialysis (HD) [3 trials, n = 154 participants; weighted mean difference (WMD) = 1.02 kg; 95% CI: 0.38, 1.65 kg; P = 0.002; I2 = 11.4%] and decreased body weight in subjects who are overweight/obese but otherwise healthy (5 trials, n = 245 participants; WMD = -0.55 kg; 95% CI: -1.06, -0.04 kg; P = 0.03; I2 = 31.5%). Dose-response analyses revealed a significant nonlinear effect of supplementation dosage on BMI (P = 0.001). Our data suggest that zinc supplementation increases body weight in patients undergoing HD and decreases body weight in individuals who are overweight/obese but otherwise healthy, although after normalization for study duration, the association observed in subjects who are overweight/obese disappeared. Although more high-quality studies are needed to reach a definitive conclusion, our study supports the view that zinc may be associated with body weight.
Collapse
Affiliation(s)
- Shima Abdollahi
- Department of Nutrition and Public Health, School of Public Health, North Khorasan University of Medical Sciences, Bojnurd, Iran
- Department of Nutrition, Faculty of Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Omid Toupchian
- Department of Nutrition and Public Health, School of Public Health, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Ahmad Jayedi
- Food Safety Research Center (Salt), Semnan University of Medical Sciences, Semnan, Iran
| | - David Meyre
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Vivian Tam
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada
| | - Sepideh Soltani
- Department of Nutrition, Faculty of Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Yazd Cardiovascular Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
25
|
黄 佳, 贾 如, 魏 晓, 罗 肖. [Time-sequential expression of lnc AK079912 during adipose tissue development and browning in mice]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2019; 39:1494-1499. [PMID: 31907161 PMCID: PMC6942996 DOI: 10.12122/j.issn.1673-4254.2019.12.15] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Indexed: 02/04/2023]
Abstract
OBJECTIVE To investigate the time-sequential expression of a novel long non-coding RNA, lnc AK079912, in metabolically related tissues and during adipose tissue development and browning in mice. METHODS The interscapular brown adipose tissue (iBAT), subcutaneous white adipose tissue (sWAT), epididymal white adipose tissue (eWAT), liver tissues and muscular tissues were collected from 8-week-old C57BL/6J mice. The iBAT, sWAT and eWAT were also collected from the mice during development (0 day, 21 days, 8 weeks and 6 months after birth) and from 8- to 10-week- mice with cold exposure (4 ℃) and intraperitoneal injections of CL316, 243 (1 μg/g body weight) for 1 to 5 days. Trizol was used to extract the total RNA from the tissues, and RT-qPCR was performed to detect the expressions of lnc AK079912. Isolated mouse preadipocytes in primary culture were induced for adipogenic differentiation for 9 days and then treated with CL316, 243 (2 μmol/L) for different durations (no longer than 24 h); the expression of lnc AK079912 in the cells was detected using RT-qPCR at different time points of the treatment. RESULTS Lnc AK079912 was highly expressed in mouse adipose tissues, the highest in iBAT, followed by the muscular tissue, but was hardly detected in the liver tissue. The expression level of lnc AK079912 increased progressively in iBAT and sWAT during development of the mice, while its expression in eWAT showed an initial increase followed by a reduction at 8 weeks (P < 0.001). No significant difference was found in the expression of lnc AK079912 in the iBAT, sWAT or eWAT in mice with cold stimulation for 1 to 5 days (P > 0.05). The expression of lnc AK079912 was significantly decreased in iBAT and eWAT (P < 0.05) but increased in eWAT from mice with intraperitoneal injection of CL316, 243 for 1 to 5 days (P < 0.05). The expression level in the adipocytes in primary culture was significantly increased in response to treatment with CL316, 243 (P < 0.05). CONCLUSIONS Lnc AK079912 is highly expressed in mouse adipose tissue, and its expression gradually increases with the development of adipose tissue but with a depot-specific difference. Lnc AK079912 is significantly elevated in the early stage of adipose tissue browning, indicating its important role in the development and browning of adipose tissue.
Collapse
Affiliation(s)
- 佳琪 黄
- 西安交通大学医学部基础医学院生理学与病理生理学系,教育部环境与疾病相关基因重点实验室,陕西 西安 710061Department of Physiology and Pathophysiology, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University School of Medicine, Xi'an 710061, China
| | - 如 贾
- 西安交通大学医学部附属口腔医院修复科,陕西 西安 710004Department of Prosthodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, China
- 陕西省颅颌面精准医学研究重点实验室,陕西 西安 710004Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, Xi'an Jiaotong University School of Medicine, Xi'an 710061, China
| | - 晓静 魏
- 西安交通大学医学部基础医学院生理学与病理生理学系,教育部环境与疾病相关基因重点实验室,陕西 西安 710061Department of Physiology and Pathophysiology, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University School of Medicine, Xi'an 710061, China
| | - 肖 罗
- 西安交通大学医学部基础医学院生理学与病理生理学系,教育部环境与疾病相关基因重点实验室,陕西 西安 710061Department of Physiology and Pathophysiology, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University School of Medicine, Xi'an 710061, China
| |
Collapse
|
26
|
Ohashi W, Hara T, Takagishi T, Hase K, Fukada T. Maintenance of Intestinal Epithelial Homeostasis by Zinc Transporters. Dig Dis Sci 2019; 64:2404-2415. [PMID: 30830525 DOI: 10.1007/s10620-019-05561-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 02/22/2019] [Indexed: 12/12/2022]
Abstract
Zinc is an essential micronutrient for normal organ function, and dysregulation of zinc metabolism has been implicated in a wide range of diseases. Emerging evidence has revealed that zinc transporters play diverse roles in cellular homeostasis and function by regulating zinc trafficking via organelles or the plasma membrane. In the gastrointestinal tract, zinc deficiency leads to diarrhea and dysfunction of intestinal epithelial cells. Studies also showed that zinc transporters are very important in intestinal epithelial homeostasis. In this review, we describe the physiological roles of zinc transporters in intestinal epithelial functions and relevance of zinc transporters in gastrointestinal diseases.
Collapse
Affiliation(s)
- Wakana Ohashi
- Department of Molecular and Medical Pharmacology, Graduate School of Medicine and Pharmaceutical Sciences for Research, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Takafumi Hara
- Molecular and Cellular Physiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Nishihamabouji, Yamashiro, Tokushima, 770-8055, Japan
| | - Teruhisa Takagishi
- Molecular and Cellular Physiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Nishihamabouji, Yamashiro, Tokushima, 770-8055, Japan
| | - Koji Hase
- Division of Biochemistry, Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo, 105-8512, Japan
| | - Toshiyuki Fukada
- Molecular and Cellular Physiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Nishihamabouji, Yamashiro, Tokushima, 770-8055, Japan.
- Division of Pathology, Department of Oral Diagnostic Sciences, School of dentistry, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan.
- RIKEN Center for Integrative Medical Sciences, 1-7-22, Suehiro-cho, Tsurumi-ku, Yokohama City, Kanagawa, 230-0042, Japan.
| |
Collapse
|
27
|
The Zinc Transporter Zip7 Is Downregulated in Skeletal Muscle of Insulin-Resistant Cells and in Mice Fed a High-Fat Diet. Cells 2019; 8:cells8070663. [PMID: 31266232 PMCID: PMC6678147 DOI: 10.3390/cells8070663] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 06/27/2019] [Accepted: 06/28/2019] [Indexed: 12/14/2022] Open
Abstract
Background: The zinc transporter Zip7 modulates zinc flux and controls cell signaling molecules associated with glucose metabolism in skeletal muscle. The present study evaluated the role of Zip7 in cell signaling pathways involved in insulin-resistant skeletal muscle and mice fed a high-fat diet. Methods: Insulin-resistant skeletal muscle cells were prepared by treatment with an inhibitor of the insulin receptor, HNMPA-(AM)3 or palmitate, and Zip7 was analyzed along with pAkt, pTyrosine and Glut4. Similarly, mice fed normal chow (NC) or a high-fat diet (HFD) were also analyzed for protein expression of Glut4 and Zip7. An overexpression system for Zip7 was utilized to determine the action of this zinc transporter on several genes implicated in insulin signaling and glucose control. Results: We identified that Zip7 is upregulated by glucose in normal skeletal muscle cells and downregulated in insulin-resistant skeletal muscle. We also observed (as expected) a decrease in pAkt and Glut4 in the insulin-resistant skeletal muscle cells. The overexpression of Zip7 in skeletal muscle cells led to the modulation of key genes involved in the insulin signaling axis and glucose metabolism including Akt3, Dok2, Fos, Hras, Kras, Nos2, Pck2, and Pparg. In an in vivo mouse model, we identified a reduction in Glut4 and Zip7 in the skeletal muscle of mice fed a HFD compared to NC controls. Conclusions: These data suggest that Zip7 plays a role in skeletal muscle insulin signaling and is downregulated in an insulin-resistant, and HFD state. Understanding the molecular mechanisms of Zip7 action will provide novel opportunities to target this transporter therapeutically for the treatment of insulin resistance and type 2 diabetes.
Collapse
|
28
|
Zhu Q, Glazier BJ, Hinkel BC, Cao J, Liu L, Liang C, Shi H. Neuroendocrine Regulation of Energy Metabolism Involving Different Types of Adipose Tissues. Int J Mol Sci 2019; 20:E2707. [PMID: 31159462 PMCID: PMC6600468 DOI: 10.3390/ijms20112707] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 05/24/2019] [Accepted: 05/29/2019] [Indexed: 12/17/2022] Open
Abstract
Despite tremendous research efforts to identify regulatory factors that control energy metabolism, the prevalence of obesity has been continuously rising, with nearly 40% of US adults being obese. Interactions between secretory factors from adipose tissues and the nervous system innervating adipose tissues play key roles in maintaining energy metabolism and promoting survival in response to metabolic challenges. It is currently accepted that there are three types of adipose tissues, white (WAT), brown (BAT), and beige (BeAT), all of which play essential roles in maintaining energy homeostasis. WAT mainly stores energy under positive energy balance, while it releases fuels under negative energy balance. Thermogenic BAT and BeAT dissipate energy as heat under cold exposure to maintain body temperature. Adipose tissues require neural and endocrine communication with the brain. A number of WAT adipokines and BAT batokines interact with the neural circuits extending from the brain to cooperatively regulate whole-body lipid metabolism and energy homeostasis. We review neuroanatomical, histological, genetic, and pharmacological studies in neuroendocrine regulation of adipose function, including lipid storage and mobilization of WAT, non-shivering thermogenesis of BAT, and browning of BeAT. Recent whole-tissue imaging and transcriptome analysis of differential gene expression in WAT and BAT yield promising findings to better understand the interaction between secretory factors and neural circuits, which represents a novel opportunity to tackle obesity.
Collapse
Affiliation(s)
- Qi Zhu
- Program of Physiology and Neuroscience, Department of Biology, Miami University, Oxford, OH 45056, USA.
| | - Bradley J Glazier
- Program of Physiology and Neuroscience, Department of Biology, Miami University, Oxford, OH 45056, USA.
| | - Benjamin C Hinkel
- Program of Physiology and Neuroscience, Department of Biology, Miami University, Oxford, OH 45056, USA.
| | - Jingyi Cao
- Program of Physiology and Neuroscience, Department of Biology, Miami University, Oxford, OH 45056, USA.
| | - Lin Liu
- Program of Bioinformatics, Department of Biology, Miami University, Oxford, OH 45056, USA.
| | - Chun Liang
- Program of Bioinformatics, Department of Biology, Miami University, Oxford, OH 45056, USA.
| | - Haifei Shi
- Program of Physiology and Neuroscience, Department of Biology, Miami University, Oxford, OH 45056, USA.
| |
Collapse
|
29
|
Zhang T, Deng B, Zhang R, Qin X, Zhang J, Zhao J. Dietary Sea Buckthorn Pomace Induces Beige Adipocyte Formation in Inguinal White Adipose Tissue in Lambs. Animals (Basel) 2019; 9:ani9040193. [PMID: 31022943 PMCID: PMC6523461 DOI: 10.3390/ani9040193] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 04/14/2019] [Accepted: 04/17/2019] [Indexed: 12/20/2022] Open
Abstract
The sea buckthorn contains substantial amounts of bioactive compounds. The objective of this study was to investigate the effects of dietary sea buckthorn pomace (SBP) on sheep beige adipocyte formation. A total of thirty lambs were equally divided into three groups and fed with diets containing different levels of SBP: 0% SBP (Control), 7.8% SBP (7.8SBP), and 16.0% SBP (16SBP). The results showed that dietary SBP affected inguinal adipocytes' size distribution, and increased both UCP1 protein content (p < 0.05) and mitochondrial numbers (p < 0.05). mRNA expression of peroxisome proliferator-activated receptor gamma coactivator 1α (PGC-1α), nuclear respiratory factor 1, and mitochondrial transcription factor A were increased when animals were subjected to 16% SBP (p < 0.05). Supplementation with 16% SBP increased CCAAT/enhancer-binding protein β content (p < 0.05) and PR domain containing 16 mRNA abundance (p < 0.05). Consistently, inguinal white adipose tissue (iWAT) from the 16SBP group exhibited increased insulin sensitivity, which was associated with elevated glucose transporter 4 abundance (p < 0.05). Importantly, AMP-activated protein kinase (AMPK) was activated in the 16SBP group (p < 0.05). Collectively, these results suggest that dietary SBP promotes iWAT browning in lambs, which might be through the activation of the AMPK-PGC-1α-UCP1 signaling pathway.
Collapse
Affiliation(s)
- Ting Zhang
- Department of Animal Sciences and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, China.
| | - Buhao Deng
- Department of Animal Sciences and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, China.
| | - Ruixin Zhang
- Department of Animal Sciences and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, China.
| | - Xuze Qin
- Department of Animal Sciences and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, China.
| | - Jianxin Zhang
- Department of Animal Sciences and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, China.
| | - Junxing Zhao
- Department of Animal Sciences and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, China.
| |
Collapse
|
30
|
Takagishi T, Hara T, Fukada T. [Investigation of the importance of zinc-signaling: insights from animal model study and human disease]. Nihon Yakurigaku Zasshi 2019; 154:327-334. [PMID: 31787685 DOI: 10.1254/fpj.154.327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Zinc (Zn) is one of the essential trace elements required for human developments and it plays an important role in the maintenance of numerous tissue homeostasis. The amount of Zn levels was below the constant level which induced the various harmful health effects such as impaired growth, hair loss, taste disturbance, anorexia. Maintenance of Zn homeostasis in body mainly depends on two families of Zn transporters; Zrt- and Irt-like proteins (ZIPs), and Zinc transporters (ZnTs). Some studies based on the gene knock-out mice and human genetic analysis have been reported the relationship between zinc transporters and human diseases. Recent studies have shown that Zn transporter-mediated Zn ion behaves as a signaling factor, called Zn signal, that exerts a multiple function in cellular events. In this review article we describe important physiological roles of Zn transporters and their contribution at the molecular, biochemical, and genetic levels underlying the mechanisms of human diseases.
Collapse
Affiliation(s)
- Teruhisa Takagishi
- Molecular and Cellular Physiology Faculty of Pharmaceutical Sciences Tokushima Bunri University
| | - Takafumi Hara
- Molecular and Cellular Physiology Faculty of Pharmaceutical Sciences Tokushima Bunri University
| | - Toshiyuki Fukada
- Molecular and Cellular Physiology Faculty of Pharmaceutical Sciences Tokushima Bunri University
| |
Collapse
|
31
|
Abstract
During the last decades, research on adipose tissues has spread in parallel with the extension of obesity. Several observations converged on the idea that adipose tissues are organized in a large organ with endocrine and plastic properties. Two parenchymal components: white (WATs) and brown adipose tissues (BATs) are contained in subcutaneous and visceral compartments. Although both have endocrine properties, their function differs: WAT store lipids to allow intervals between meals, BAT burns lipids for thermogenesis. In spite of these opposite functions, they share the ability for reciprocal reversible transdifferentiation to tackle special physiologic needs. Thus, chronic need for thermogenesis induces browning and chronic positive energy balance induce whitening. Lineage tracing and data from explant studies strongly suggest other remodeling properties of this organ. During pregnancy and lactation breast WAT transdifferentiates into milk-secreting glands, composed by cells with abundant cytoplasmic lipids (pink adipocytes) and in the postlactation period pink adipocytes transdifferentiate back into WAT and BAT. The plastic properties of mature adipocytes are supported also by a liposecretion process in vitro where adult cell in culture transdifferentiate to differentiated fibroblast-like elements able to give rise to different phenotypes (rainbow adipocytes). In addition, the inflammasome system is activated in stressed adipocytes from obese adipose tissue. These adipocytes die and debris are reabsorbed by macrophages inducing a chronic low-grade inflammation, potentially contributing to insulin resistance and T2 diabetes. Thus, the plastic properties of this organ could open new therapeutic perspectives in the obesity-related metabolic disease and in breast pathologies. © 2018 American Physiological Society. Compr Physiol 8:1357-1431, 2018.
Collapse
Affiliation(s)
- Saverio Cinti
- Professor of Human Anatomy, Director, Center of Obesity, University of Ancona (Politecnica delle Marche), Ancona, Italy
| |
Collapse
|
32
|
Insulin promoter in human pancreatic β cells contacts diabetes susceptibility loci and regulates genes affecting insulin metabolism. Proc Natl Acad Sci U S A 2018; 115:E4633-E4641. [PMID: 29712868 DOI: 10.1073/pnas.1803146115] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Both type 1 and type 2 diabetes involve a complex interplay between genetic, epigenetic, and environmental factors. Our laboratory has been interested in the physical interactions, in nuclei of human pancreatic β cells, between the insulin (INS) gene and other genes that are involved in insulin metabolism. We have identified, using Circularized Chromosome Conformation Capture (4C), many physical contacts in a human pancreatic β cell line between the INS promoter on chromosome 11 and sites on most other chromosomes. Many of these contacts are associated with type 1 or type 2 diabetes susceptibility loci. To determine whether physical contact is correlated with an ability of the INS locus to affect expression of these genes, we knock down INS expression by targeting the promoter; 259 genes are either up or down-regulated. Of these, 46 make physical contact with INS We analyze a subset of the contacted genes and show that all are associated with acetylation of histone H3 lysine 27, a marker of actively expressed genes. To demonstrate the usefulness of this approach in revealing regulatory pathways, we identify from among the contacted sites the previously uncharacterized gene SSTR5-AS1 and show that it plays an important role in controlling the effect of somatostatin-28 on insulin secretion. These results are consistent with models in which clustering of genes supports transcriptional activity. This may be a particularly important mechanism in pancreatic β cells and in other cells where a small subset of genes is expressed at high levels.
Collapse
|
33
|
The Role of the Slc39a Family of Zinc Transporters in Zinc Homeostasis in Skin. Nutrients 2018; 10:nu10020219. [PMID: 29462920 PMCID: PMC5852795 DOI: 10.3390/nu10020219] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 01/27/2018] [Accepted: 02/11/2018] [Indexed: 12/20/2022] Open
Abstract
The first manifestations that appear under zinc deficiency are skin defects such as dermatitis, alopecia, acne, eczema, dry, and scaling skin. Several genetic disorders including acrodermatitis enteropathica (also known as Danbolt-Closs syndrome) and Brandt's syndrome are highly related to zinc deficiency. However, the zinc-related molecular mechanisms underlying normal skin development and homeostasis, as well as the mechanism by which disturbed zinc homeostasis causes such skin disorders, are unknown. Recent genomic approaches have revealed the physiological importance of zinc transporters in skin formation and clarified their functional impairment in cutaneous pathogenesis. In this review, we provide an overview of the relationships between zinc deficiency and skin disorders, focusing on the roles of zinc transporters in the skin. We also discuss therapeutic outlooks and advantages of controlling zinc levels via zinc transporters to prevent cutaneous disorganization.
Collapse
|
34
|
Abstract
The skin is the third most zinc (Zn)-abundant tissue in the body. The skin consists of the epidermis, dermis, and subcutaneous tissue, and each fraction is composed of various types of cells. Firstly, we review the physiological functions of Zn and Zn transporters in these cells. Several human disorders accompanied with skin manifestations are caused by mutations or dysregulation in Zn transporters; acrodermatitis enteropathica (Zrt-, Irt-like protein (ZIP)4 in the intestinal epithelium and possibly epidermal basal keratinocytes), the spondylocheiro dysplastic form of Ehlers-Danlos syndrome (ZIP13 in the dermal fibroblasts), transient neonatal Zn deficiency (Zn transporter (ZnT)2 in the secretory vesicles of mammary glands), and epidermodysplasia verruciformis (ZnT1 in the epidermal keratinocytes). Additionally, acquired Zn deficiency is deeply involved in the development of some diseases related to nutritional deficiencies (acquired acrodermatitis enteropathica, necrolytic migratory erythema, pellagra, and biotin deficiency), alopecia, and delayed wound healing. Therefore, it is important to associate the existence of mutations or dysregulation in Zn transporters and Zn deficiency with skin manifestations.
Collapse
|
35
|
Role of Zinc Homeostasis in the Pathogenesis of Diabetes and Obesity. Int J Mol Sci 2018; 19:ijms19020476. [PMID: 29415457 PMCID: PMC5855698 DOI: 10.3390/ijms19020476] [Citation(s) in RCA: 160] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 01/30/2018] [Accepted: 02/02/2018] [Indexed: 12/11/2022] Open
Abstract
Zinc deficiency is a risk factor for obesity and diabetes. However, until recently, the underlying molecular mechanisms remained unclear. The breakthrough discovery that the common polymorphism in zinc transporter SLC30A8/ZnT8 may increase susceptibility to type 2 diabetes provided novel insights into the role of zinc in diabetes. Our group and others showed that altered ZnT8 function may be involved in the pathogenesis of type 2 diabetes, indicating that the precise control of zinc homeostasis is crucial for maintaining health and preventing various diseases, including lifestyle-associated diseases. Recently, the role of the zinc transporter ZIP13 in the regulation of beige adipocyte biogenesis was clarified, which indicated zinc homeostasis regulation as a possible therapeutic target for obesity and metabolic syndrome. Here we review advances in the role of zinc homeostasis in the pathophysiology of diabetes, and propose that inadequate zinc distribution may affect the onset of diabetes and metabolic diseases by regulating various critical biological events.
Collapse
|
36
|
Olechnowicz J, Tinkov A, Skalny A, Suliburska J. Zinc status is associated with inflammation, oxidative stress, lipid, and glucose metabolism. J Physiol Sci 2018; 68:19-31. [PMID: 28965330 PMCID: PMC5754376 DOI: 10.1007/s12576-017-0571-7] [Citation(s) in RCA: 345] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 09/18/2017] [Indexed: 12/14/2022]
Abstract
A number of studies have reported that zinc plays a substantial role in the development of metabolic syndrome, taking part in the regulation of cytokine expression, suppressing inflammation, and is also required to activate antioxidant enzymes that scavenge reactive oxygen species, reducing oxidative stress. Zinc also plays a role in the correct functioning of lipid and glucose metabolism, regulating and forming the expression of insulin. In numerous studies, zinc supplementation has been found to improve blood pressure, glucose, and LDL cholesterol serum level. Deeper knowledge of zinc's properties may help in treating metabolic syndrome, thus protecting against stroke and angina pectoris, and ultimately against death.
Collapse
Affiliation(s)
- J Olechnowicz
- Poznan University of Life Sciences, ul. Wojska Polskiego 31, 62-624, Poznan , Poland
| | - A Tinkov
- Orenburg State Medical University, Sovetskaya St., 6, Orenburg, 460000, Russia
- Orenburg State University, Pobedy Avenue, 13, Orenburg, 460018, Russia
- RUDN University, Miklukho-Maklay St., 10/2, Moscow, 117198, Russia
- Yaroslavl State University, Sovetskaya St., 14, Yaroslavl, 150000, Russia
| | - A Skalny
- Orenburg State University, Pobedy Avenue, 13, Orenburg, 460018, Russia
- RUDN University, Miklukho-Maklay St., 10/2, Moscow, 117198, Russia
- Yaroslavl State University, Sovetskaya St., 14, Yaroslavl, 150000, Russia
- All-Russian Research Institute of Medicinal and Aromatic Plants (VILAR), Grina St., 7, Moscow, 117216, Russia
| | - Joanna Suliburska
- Poznan University of Life Sciences, ul. Wojska Polskiego 31, 62-624, Poznan , Poland.
| |
Collapse
|
37
|
Recent Advances in the Role of SLC39A/ZIP Zinc Transporters In Vivo. Int J Mol Sci 2017; 18:ijms18122708. [PMID: 29236063 PMCID: PMC5751309 DOI: 10.3390/ijms18122708] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 11/27/2017] [Accepted: 12/08/2017] [Indexed: 02/07/2023] Open
Abstract
Zinc (Zn), which is an essential trace element, is involved in numerous mammalian physiological events; therefore, either a deficiency or excess of Zn impairs cellular machineries and influences physiological events, such as systemic growth, bone homeostasis, skin formation, immune responses, endocrine function, and neuronal function. Zn transporters are thought to mainly contribute to Zn homeostasis within cells and in the whole body. Recent genetic, cellular, and molecular studies of Zn transporters highlight the dynamic role of Zn as a signaling mediator linking several cellular events and signaling pathways. Dysfunction in Zn transporters causes various diseases. This review aims to provide an update of Zn transporters and Zn signaling studies and discusses the remaining questions and future directions by focusing on recent progress in determining the roles of SLC39A/ZIP family members in vivo.
Collapse
|
38
|
Norouzi S, Adulcikas J, Sohal SS, Myers S. Zinc transporters and insulin resistance: therapeutic implications for type 2 diabetes and metabolic disease. J Biomed Sci 2017; 24:87. [PMID: 29157234 PMCID: PMC5694903 DOI: 10.1186/s12929-017-0394-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 11/14/2017] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Zinc is a metal ion that is essential for growth and development, immunity, and metabolism, and therefore vital for life. Recent studies have highlighted zinc's dynamic role as an insulin mimetic and a cellular second messenger that controls many processes associated with insulin signaling and other downstream pathways that are amendable to glycemic control. MAIN BODY Mechanisms that contribute to the decompartmentalization of zinc and dysfunctional zinc transporter mechanisms, including zinc signaling are associated with metabolic disease, including type 2 diabetes. The actions of the proteins involved in the uptake, storage, compartmentalization and distribution of zinc in cells is under intense investigation. Of these, emerging research has highlighted a role for several zinc transporters in the initiation of zinc signaling events in cells that lead to metabolic processes associated with maintaining insulin sensitivity and thus glycemic homeostasis. CONCLUSION This raises the possibility that zinc transporters could provide novel utility to be targeted experimentally and in a clinical setting to treat patients with insulin resistance and thus introduce a new class of drug target with utility for diabetes pharmacotherapy.
Collapse
Affiliation(s)
- Shaghayegh Norouzi
- Faculty of Health, School of Health Sciences, University of Tasmania, Newnham Campus, Launceston, TAS, 7250, Australia
| | - John Adulcikas
- Faculty of Health, School of Health Sciences, University of Tasmania, Newnham Campus, Launceston, TAS, 7250, Australia
| | - Sukhwinder Singh Sohal
- Faculty of Health, School of Health Sciences, University of Tasmania, Newnham Campus, Launceston, TAS, 7250, Australia
| | - Stephen Myers
- Faculty of Health, School of Health Sciences, University of Tasmania, Newnham Campus, Launceston, TAS, 7250, Australia.
| |
Collapse
|
39
|
Hojyo S, Bin BH, Fukada T. Dysregulated zinc homeostasis in rare skin disorders. Expert Opin Orphan Drugs 2017. [DOI: 10.1080/21678707.2017.1394184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Shintaro Hojyo
- Osteoimmunology, Deutsches Rheuma-Forschungszentrum, Berlin, Berlin, Germany
| | - Bum-Ho Bin
- Bioscience Research Institute, Amorepacific Corporation R&D Center, Yongin-si, Republic of Korea
| | - Toshiyuki Fukada
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, Japan
| |
Collapse
|